1
|
Chen Z, Kwek JL, Lim RS, Yong YR, Loh AHL, Lim WK, Teo JX, Tay KSS, Ng PS. Long term follow-up of multiorgan disease in Kleefstra syndrome 2 in an adult - case report. BMC Neurol 2025; 25:199. [PMID: 40329253 PMCID: PMC12057049 DOI: 10.1186/s12883-025-04210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVES The Kleefstra syndrome spectrum (KSS) is a group of neurodevelopmental disorders characterized by intellectual disability, behavioral disorders, growth and neurodevelopmental delay, facial dysmorphism and neurological deficits. Kleefstra syndrome 2 (KLEFS2) is a part of KSS and is due to heterozygous loss-of-function variants in the KMT2 C gene. We report the long-term clinical course and multi-organ manifestations of a patient with KLEFS2 caused by a novel heterozygous pathogenic variant in KMT2 C. METHODS A patient with KSS phenotype developed proteinuria with progressive kidney dysfunction secondary to focal segmental glomerular sclerosis. She subsequently developed recurrent episodes that mimicked mitochondrial stroke-like episodes. The phenotype included encephalopathy, stroke-like episodes with focal status epilepticus with impaired consciousness associated with cortical and subcortical T2/FLAIR signal hyperintensities that partially responded to intravenous arginine infusions. RESULTS Exome sequencing revealed a heterozygous pathogenic nonsense variant in KMT2 C (NM_170606.3) c.3940C > T (p.Gln1314Ter). Nuclear and mitochondrial DNA variants associated with mitochondrial disorders have been excluded. DISCUSSION This is a case of KLEFS2 with longitudinal 10 year follow up and its previously unreported multi-organ clinical manifestations including stroke-like episodes and nephrotic disease. Our report further expands the phenotypic spectrum of KLEFS2. Further reports of patients with KLEFS2 with multi-organ involvement should be sought to confirm our findings.
Collapse
Affiliation(s)
- Zhiyong Chen
- Department of Neurology, National Neuroscience Institute (Tan Tock Seng Hospital Campus), 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore.
| | - Jia Liang Kwek
- Department of Renal Medicine, Singapore General Hospital, Singapore, Singapore
| | - Ru Sin Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Yan Rong Yong
- Department of Diagnostic Radiology, Changi General Hospital, Singapore, Singapore
| | - Alwin Hwai Liang Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
| | - Jing Xian Teo
- SingHealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore
| | - Karine Su Shan Tay
- Department of Neurology, National Neuroscience Institute (Tan Tock Seng Hospital Campus), 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| | - Peng Soon Ng
- Department of Neurology, National Neuroscience Institute (Tan Tock Seng Hospital Campus), 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
| |
Collapse
|
2
|
Abdelwahab O, Torkamaneh D. Artificial intelligence in variant calling: a review. FRONTIERS IN BIOINFORMATICS 2025; 5:1574359. [PMID: 40337525 PMCID: PMC12055765 DOI: 10.3389/fbinf.2025.1574359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Artificial intelligence (AI) has revolutionized numerous fields, including genomics, where it has significantly impacted variant calling, a crucial process in genomic analysis. Variant calling involves the detection of genetic variants such as single nucleotide polymorphisms (SNPs), insertions/deletions (InDels), and structural variants from high-throughput sequencing data. Traditionally, statistical approaches have dominated this task, but the advent of AI led to the development of sophisticated tools that promise higher accuracy, efficiency, and scalability. This review explores the state-of-the-art AI-based variant calling tools, including DeepVariant, DNAscope, DeepTrio, Clair, Clairvoyante, Medaka, and HELLO. We discuss their underlying methodologies, strengths, limitations, and performance metrics across different sequencing technologies, alongside their computational requirements, focusing primarily on SNP and InDel detection. By comparing these AI-driven techniques with conventional methods, we highlight the transformative advancements AI has introduced and its potential to further enhance genomic research.
Collapse
Affiliation(s)
- Omar Abdelwahab
- Département de Phytologie, Université Laval, Québec City, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec City, QC, Canada
- Centre de recherche et d’innovation sur les végétaux (CRIV), Université Laval, Québec City, QC, Canada
- Institut intelligence et données (IID), Université Laval, Québec City, QC, Canada
| | - Davoud Torkamaneh
- Département de Phytologie, Université Laval, Québec City, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec City, QC, Canada
- Centre de recherche et d’innovation sur les végétaux (CRIV), Université Laval, Québec City, QC, Canada
- Institut intelligence et données (IID), Université Laval, Québec City, QC, Canada
| |
Collapse
|
3
|
Kim JA, Jang SH, Joo SY, Kim SJ, Choi JY, Jung J, Gee HY. Systematic genetic assessment of hearing loss using whole-genome sequencing identifies pathogenic variants. Exp Mol Med 2025; 57:775-787. [PMID: 40164689 PMCID: PMC12046045 DOI: 10.1038/s12276-025-01428-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 04/02/2025] Open
Abstract
Hearing loss is a clinically and genetically heterogeneous sensorineural disease that affects approximately 1 out of 1000 newborns. For the molecular diagnosis of genetic hearing loss, target panel or whole-exome sequencing (WES) have been widely used due to their cost-effectiveness and efficacy. Despite the advantages of WES, the plausible diagnoses in a substantial number of patients remain elusive due to its limited coverage. Here we utilized whole-genome sequencing (WGS) on a large cohort of individuals with hearing loss to overcome the drawbacks of WES and find the advantages of WGS. We implemented a systematic workflow to identify coding region variants, cryptic splice variants, mitochondrial variants, copy number variants, cis-regulatory variants and transposable element insertions. WGS was conducted on 140 families with hearing loss. Causative variations were identified in 37 of these families, accounting for 26% of the total. WGS possessed the capability to find genetic variations that are not identifiable using WES. The identified variants by WGS in this study encompassed aberrant splicing variants in EYA1 and CDH23, mitochondrial variants in MT-RNR1 and MT-CO1, structural variants in STRC, and Alu insertion in SLC17A8. These findings highlight the benefits of WGS. With the decreasing cost of WGS, its usage will become more prevalent, allowing more precise identification of the genetic causes of hearing loss.
Collapse
Affiliation(s)
- Jung Ah Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea
| | - Seung Hyun Jang
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea
| | - Sun Yung Joo
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea
| | - Se Jin Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea
| | - Jae Young Choi
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinsei Jung
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea.
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Won-Sang Lee Institute for Hearing Loss, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Magistrati M, Zupin L, Lamantea E, Baruffini E, Ghezzi D, Legati A, Celsi F, Murru FM, Capaci V, Pinamonti M, Bussani R, Carrozzi M, Dallabona C, Zeviani M, Bonati MT. De Novo DNM1L Pathogenic Variant Associated with Lethal Encephalocardiomyopathy-Case Report and Literature Review. Int J Mol Sci 2025; 26:846. [PMID: 39859560 PMCID: PMC11765995 DOI: 10.3390/ijms26020846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pathogenic variants in DNM1L, encoding dynamin-like protein-1 (DRP1), cause a lethal encephalopathy. DRP1 defective function results in altered mitochondrial networks, characterized by elongated/spaghetti-like, highly interconnected mitochondria. We validated in yeast the pathogenicity of a de novo DNM1L variant identified by whole exome sequencing performed more than 10 years after the patient's death. Meanwhile, we reviewed the broadness and specificities of DNM1L-related phenotype. The patient, who exhibited developmental delay in her third year, developed a therapy-refractory myoclonic status epilepticus, followed by neurological deterioration with brain atrophy and refractory epilepsy. She died of heart failure due to hypertrophic cardiomyopathy. She was found to be heterozygous for the DNM1L variant (NM_ 012062.5):c.1201G>A, p.(Gly401Ser). We demonstrated its deleterious impact and dominant negative effect by assessing haploid and diploid mutant yeast strains, oxidative growth, oxygen consumption, frequency of petite, and architecture of the mitochondrial network. Structural modeling of p.(Gly401Ser) predicted the interference of the mutant protein in the self-oligomerization of the DRP1 active complex. DNM1L-related phenotypes include static or (early) lethal encephalopathy and neurodevelopmental disorders. In addition, there may be ophthalmological impairment, peripheral neuropathy, ataxia, dystonia, spasticity, myoclonus, and myopathy. The clinical presentations vary depending on mutations in different DRP1 domains. Few pathogenic variants, the p.(Gly401Ser) included, cause an encephalocardiomyopathy with refractory status epilepticus.
Collapse
Affiliation(s)
- Martina Magistrati
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, 43124 Parma, Italy; (M.M.); (C.D.)
| | - Luisa Zupin
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Eleonora Lamantea
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133 Milan, Italy; (E.L.); (D.G.); (A.L.)
| | - Enrico Baruffini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, 43124 Parma, Italy; (M.M.); (C.D.)
| | - Daniele Ghezzi
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133 Milan, Italy; (E.L.); (D.G.); (A.L.)
- Department of Pathophysiology and Transplantation, University of Milan, Via F. Sforza, 35, 20122 Milan, Italy
| | - Andrea Legati
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria, 11, 20133 Milan, Italy; (E.L.); (D.G.); (A.L.)
| | - Fulvio Celsi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Flora Maria Murru
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Valeria Capaci
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Maurizio Pinamonti
- Institute of Pathological Anatomy and Histology, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Via Giacomo Puccini, 50, 34148 Trieste, Italy; (M.P.); (R.B.)
| | - Rossana Bussani
- Institute of Pathological Anatomy and Histology, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), University of Trieste, Via Giacomo Puccini, 50, 34148 Trieste, Italy; (M.P.); (R.B.)
- Department of Medical, Surgical and Health Sciences, University of Trieste, Via Giacomo Puccini, 50, 34148 Trieste, Italy
| | - Marco Carrozzi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze, 11/A, 43124 Parma, Italy; (M.M.); (C.D.)
| | - Massimo Zeviani
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| | - Maria Teresa Bonati
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy; (L.Z.); (F.C.); (F.M.M.); (V.C.); (M.C.); (M.Z.)
| |
Collapse
|
5
|
Stenton SL, Laricchia K, Lake NJ, Chaluvadi S, Ganesh V, DiTroia S, Osei-Owusu I, Pais L, O’Heir E, Austin-Tse C, O’Leary M, Abu Shanap M, Barrows C, Berger S, Bönnemann CG, Bujakowska KM, Campagna DR, Compton AG, Donkervoort S, Fleming MD, Gallacher L, Gleeson JG, Haliloglu G, Pierce EA, Place EM, Sankaran VG, Shimamura A, Stark Z, Tan TY, Thorburn DR, White SM, Genomics Research to Elucidate the Genetics of Rare Diseases (GREGoR) Consortium, Vilain E, Lek M, Rehm HL, O’Donnell-Luria A. Mitochondrial DNA variant detection in over 6,500 rare disease families by the systematic analysis of exome and genome sequencing data resolves undiagnosed cases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.22.24319370. [PMID: 39763565 PMCID: PMC11703311 DOI: 10.1101/2024.12.22.24319370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Background Variants in the mitochondrial genome (mtDNA) cause a diverse collection of mitochondrial diseases and have extensive phenotypic overlap with Mendelian diseases encoded on the nuclear genome. The mtDNA is often not specifically evaluated in patients with suspected Mendelian disease, resulting in overlooked diagnostic variants. Methods Using dedicated pipelines to address the technical challenges posed by the mtDNA - circular genome, variant heteroplasmy, and nuclear misalignment - single nucleotide variants, small indels, and large mtDNA deletions were called from exome and genome sequencing data, in addition to RNA-sequencing when available. A cohort of 6,660 rare disease families were analyzed (5,625 genetically undiagnosed, 84%) from the Genomics Research to Elucidate the Genetics of Rare diseases (GREGoR) Consortium as well as other rare disease cohorts. Results Diagnostic mtDNA variants were identified in 10 previously genetically undiagnosed families (one large deletion, eight reported pathogenic variants, one novel pathogenic variant). In one additional undiagnosed proband, the detection of >900 heteroplasmic variants provided functional evidence of pathogenicity to a novel de novo variant in the nuclear gene POLG (DNA polymerase gamma), responsible for mtDNA replication and repair. Conclusion mtDNA variant calling from data generated by exome and genome sequencing for nuclear variant analysis resulted in a genetic diagnosis or detection of a candidate variant for 0.4% of undiagnosed families affected by a broad range of rare diseases.
Collapse
Affiliation(s)
- Sarah L. Stenton
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristen Laricchia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Nicole J. Lake
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sushma Chaluvadi
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Vijay Ganesh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Stephanie DiTroia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Ikeoluwa Osei-Owusu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Lynn Pais
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Emily O’Heir
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Christina Austin-Tse
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Melanie O’Leary
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Mayada Abu Shanap
- Hematology/Oncology, Bone Marrow Transplantation and Cellular Therapy, Pediatric Department, King Hussein Cancer Centre (KHCC), Amman, Jordan
| | - Chelsea Barrows
- University of California, Department of Neurosciences, San Diego, CA; Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Seth Berger
- Children’s National Research Institute, Washington, DC, USA
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kinga M. Bujakowska
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dean R. Campagna
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alison G. Compton
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark D. Fleming
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lyndon Gallacher
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Joseph G. Gleeson
- University of California, Department of Neurosciences, San Diego, CA; Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Goknur Haliloglu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eric A. Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Emily M. Place
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Akiko Shimamura
- Department of Hematology and Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - David R. Thorburn
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Susan M. White
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | | | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Heidi L. Rehm
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anne O’Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
6
|
Leung EYL, Robbins HL, Zaman S, Lal N, Morton D, Dew L, Williams AP, Wallis Y, Bell J, Raghavan M, Middleton G, Beggs AD. The potential clinical utility of Whole Genome Sequencing for patients with cancer: evaluation of a regional implementation of the 100,000 Genomes Project. Br J Cancer 2024; 131:1805-1813. [PMID: 39478124 PMCID: PMC11589591 DOI: 10.1038/s41416-024-02890-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND The 100,000 Genomes Project established infrastructure for Whole Genome Sequencing (WGS) in the United Kingdom. METHODS A retrospective study of cancer patients recruited to the 100,000 Genomes Project by the West Midlands Genomics Medicine Centre, evaluating clinical relevance of results. RESULTS After excluding samples with no sequencing data (1678/4851; 34.6%), 3166 sample sets (germline and somatic) from 3067 participants were sequenced. Results of 1256 participants (41.0%) were interpreted (excluding participants who died (308/3067; 10.0%) or were clinically excluded (1503/3067; 49.0%)). Of these, 323 (25.7%) had no variants in genes which may alter management (Domain 1 genes). Of the remaining 933 participants, 552 (59.2%) had clinical recommendations made (718 recommendations in total). These included therapeutic recommendations (377/933; 40.4%), such as clinical trial, unlicensed or licensed therapies or high TMB recommendations, and germline variants warranting clinical genetics review (85/933; 9.1%). At the last follow up, 20.2% of all recommendations were followed (145/718). However, only a small proportion of therapeutic recommendations were followed (5.1%, 25/491). CONCLUSIONS The 100,000 Genomes Project has established infrastructure and regional experience to support personalised cancer care. The majority of those with successful sequencing had actionable variants. Ensuring GTAB recommendations are followed will maximise benefits for patients.
Collapse
Affiliation(s)
- Elaine Y L Leung
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Helen L Robbins
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Shafquat Zaman
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Neeraj Lal
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Dion Morton
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Lisa Dew
- Central and South Genomic Medicine Service Alliance, Birmingham, UK
| | - Anthony P Williams
- The Wessex NHS Genomics Medicine Centre (WGMC), the University of Southampton, Southampton, UK
| | - Yvonne Wallis
- The West Midlands Regional Genomics Laboratory (WMRGL), Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Jennie Bell
- The West Midlands Regional Genomics Laboratory (WMRGL), Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Manoj Raghavan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
7
|
Josephs KS, Seaby EG, May P, Theotokis P, Yu J, Andreou A, Sinclair H, Morris-Rosendahl D, Thomas ERA, Ennis S, Roberts AM, Ware JS. Cardiomyopathies in 100,000 genomes project: interval evaluation improves diagnostic yield and informs strategies for ongoing gene discovery. Genome Med 2024; 16:125. [PMID: 39472908 PMCID: PMC11520845 DOI: 10.1186/s13073-024-01390-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 09/24/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Cardiomyopathies are clinically important conditions, with a strong genetic component. National genomic initiatives such as 100,000 Genome Project (100KGP) provide opportunity to study these rare conditions at scale beyond conventional research studies. METHODS We present the clinical and molecular characteristics of the 100KGP cohort, comparing paediatric and adult probands with diverse cardiomyopathies. We assessed the diagnostic yield and spectrum of genetic aetiologies across clinical presentations. We re-analysed existing genomic data using an updated analytical strategy (revised gene panels; unbiased analyses of de novo variants; and improved variant prioritisation strategies) to identify new causative variants in genetically unsolved children. RESULTS We identified 1918 individuals (1563 probands, 355 relatives) with cardiomyopathy (CM) in 100KGP. Probands, comprising 273 children and 1290 adults, were enrolled under > 55 different recruitment categories. Paediatric probands had higher rates of co-existing congenital heart disease (12%) compared to adults (0.9%). Diagnostic yield following 100KGP's initial analysis was significantly higher for children (19%) than for adults (11%) with 11% of diagnoses overall made in genes not on the existing UK paediatric or syndromic CM panel. Our re-analysis of paediatric probands yields a potential diagnosis in 40%, identifying new probable or possible diagnoses in 49 previously unsolved paediatric cases. Structural and intronic variants accounted for 11% of all potential diagnoses in children while de novo variants were identified in 17%. CONCLUSIONS 100KGP demonstrates the benefit of genome sequencing over a standalone panel in CM. Re-analysis of paediatric CM probands allowed a significant uplift in diagnostic yield, emphasising the importance of iterative re-evaluation in genomic studies. Despite these efforts, many children with CM remain without a genetic diagnosis, highlighting the need for better gene-disease relationship curation and ongoing data sharing. The 100KGP CM cohort is likely to be useful for further gene discovery, but heterogeneous ascertainment and key technical limitations must be understood and addressed.
Collapse
Affiliation(s)
- Katherine S Josephs
- National Heart and Lung Institute, Imperial College London, London, UK.
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Eleanor G Seaby
- Genomic Informatics Group, Faculty of Medicine, University of Southampton, Southampton, UK
- Paediatric Infectious Diseases, Imperial College London, St Mary's Hospital, London, UK
- Translational Genomics Group, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Philippa May
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, UK
| | - Pantazis Theotokis
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Jing Yu
- The Innovation Building, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Avgi Andreou
- SSt George's University Hospitals NHS Foundation Trust, St George's University of London, London, UK
| | | | - Deborah Morris-Rosendahl
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Sarah Ennis
- Genomic Informatics Group, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Angharad M Roberts
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| |
Collapse
|
8
|
Dashti M, Ali NM, Alsaleh H, John SE, Nizam R, Al-Mulla F, Thanaraj TA. Mitochondrial haplogroup R offers protection against obesity in Kuwaiti and Qatari populations. Front Endocrinol (Lausanne) 2024; 15:1449374. [PMID: 39464187 PMCID: PMC11502345 DOI: 10.3389/fendo.2024.1449374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/19/2024] [Indexed: 10/29/2024] Open
Abstract
Background The Kuwaiti and Qatari populations have a high prevalence of obesity, a major risk factor for various metabolic disorders. Previous studies have independently explored mitochondrial DNA (mtDNA) variations and their association with obesity in these populations. This study aims to investigate the role of mtDNA haplogroups and variants in obesity risk among these Gulf populations. Methods Whole exome sequencing data from 1,112 participants (348 Kuwaitis and 764 Qataris) were analyzed for mtDNA variants. Participants were classified as obese or non-obese based on body mass index (BMI). Association analyses were performed to examine the relationship between mtDNA haplogroups and obesity, adjusting for covariates such as age and sex. Results Haplogroup R was found to be protective against obesity, with an odds ratio (OR) of 0.69 (p = 0.045). This association remained significant after adjusting for age and sex (OR = 0.694; 95% CI: 0.482-0.997; p = 0.048). Several mtDNA variants, particularly those involved in mitochondrial energy metabolism, showed nominal associations with obesity, but these did not remain significant after correcting for multiple testing. Conclusion Haplogroup R consistently demonstrates a protective association against obesity in both Kuwaiti and Qatari populations, highlighting its potential as a biomarker for obesity risk in the Gulf region. However, further research with larger sample sizes is needed to validate these findings and clarify the role of mtDNA variants in obesity.
Collapse
Affiliation(s)
- Mohammed Dashti
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Naser M. Ali
- Department of Medical Laboratories, Ahmadi Hospital, Kuwait Oil Company (KOC), Ahmadi, Kuwait
| | - Hussain Alsaleh
- Saad Al-Abdullah Academy for Security Sciences, Ministry of Interior, Shuwaikh, Kuwait
| | - Sumi Elsa John
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | | |
Collapse
|
9
|
Rouzier C, Pion E, Chaussenot A, Bris C, Ait‐El‐Mkadem Saadi S, Desquiret‐Dumas V, Gueguen N, Fragaki K, Amati‐Bonneau P, Barcia G, Gaignard P, Steffann J, Pennisi A, Bonnefont J, Lebigot E, Bannwarth S, Francou B, Rucheton B, Sternberg D, Martin‐Negrier M, Trimouille A, Hardy G, Allouche S, Acquaviva‐Bourdain C, Pagan C, Lebre A, Reynier P, Cossee M, Attarian S, Paquis‐Flucklinger V, MitoDiag's Network Collaborators, Procaccio V. Primary mitochondrial disorders and mimics: Insights from a large French cohort. Ann Clin Transl Neurol 2024; 11:1478-1491. [PMID: 38703036 PMCID: PMC11187946 DOI: 10.1002/acn3.52062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/23/2024] [Indexed: 05/06/2024] Open
Abstract
OBJECTIVE The objective of this study was to evaluate the implementation of NGS within the French mitochondrial network, MitoDiag, from targeted gene panels to whole exome sequencing (WES) or whole genome sequencing (WGS) focusing on mitochondrial nuclear-encoded genes. METHODS Over 2000 patients suspected of Primary Mitochondrial Diseases (PMD) were sequenced by either targeted gene panels, WES or WGS within MitoDiag. We described the clinical, biochemical, and molecular data of 397 genetically confirmed patients, comprising 294 children and 103 adults, carrying pathogenic or likely pathogenic variants in nuclear-encoded genes. RESULTS The cohort exhibited a large genetic heterogeneity, with the identification of 172 distinct genes and 253 novel variants. Among children, a notable prevalence of pathogenic variants in genes associated with oxidative phosphorylation (OXPHOS) functions and mitochondrial translation was observed. In adults, pathogenic variants were primarily identified in genes linked to mtDNA maintenance. Additionally, a substantial proportion of patients (54% (42/78) and 48% (13/27) in children and adults, respectively), undergoing WES or WGS testing displayed PMD mimics, representing pathologies that clinically resemble mitochondrial diseases. INTERPRETATION We reported the largest French cohort of patients suspected of PMD with pathogenic variants in nuclear genes. We have emphasized the clinical complexity of PMD and the challenges associated with recognizing and distinguishing them from other pathologies, particularly neuromuscular disorders. We confirmed that WES/WGS, instead of panel approach, was more valuable to identify the genetic basis in patients with "possible" PMD and we provided a genetic testing flowchart to guide physicians in their diagnostic strategy.
Collapse
Affiliation(s)
- Cécile Rouzier
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | - Emmanuelle Pion
- Filnemus, laboratoire de génétique moléculaire, CHUMontpellierFrance
| | - Annabelle Chaussenot
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | - Céline Bris
- Service de génétique, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| | - Samira Ait‐El‐Mkadem Saadi
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | - Valérie Desquiret‐Dumas
- Service de biochimie et biologie moléculaire, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| | - Naïg Gueguen
- Service de biochimie et biologie moléculaire, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| | - Konstantina Fragaki
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | - Patrizia Amati‐Bonneau
- Service de biochimie et biologie moléculaire, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| | - Giulia Barcia
- Service de médecine génomique des maladies rares, Hôpital Necker‐Enfants MaladesUniversité Paris Cité, Institut Imagine Unité UMR 1161ParisFrance
| | - Pauline Gaignard
- Service de Biochimie, GHU APHP Paris SaclayHôpital BicêtreLe Kremlin‐BicêtreFrance
| | - Julie Steffann
- Service de médecine génomique des maladies rares, Hôpital Necker‐Enfants MaladesUniversité Paris Cité, Institut Imagine Unité UMR 1161ParisFrance
| | - Alessandra Pennisi
- Service de médecine génomique des maladies rares, Hôpital Necker‐Enfants MaladesUniversité Paris Cité, Institut Imagine Unité UMR 1161ParisFrance
| | - Jean‐Paul Bonnefont
- Service de médecine génomique des maladies rares, Hôpital Necker‐Enfants MaladesUniversité Paris Cité, Institut Imagine Unité UMR 1161ParisFrance
| | - Elise Lebigot
- Service de Biochimie, GHU APHP Paris SaclayHôpital BicêtreLe Kremlin‐BicêtreFrance
| | - Sylvie Bannwarth
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | - Bruno Francou
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | | | - Damien Sternberg
- Unité Fonctionnelle de cardiogénétique et myogénétique moléculaire et cellulaire, Centre de génétique moléculaire et chromosomiqueAP‐HP Sorbonne Université, Hopital de la Pitié‐SalpêtrièreParisFrance
| | - Marie‐Laure Martin‐Negrier
- Unité fonctionnelle d'histologie moléculaire, Service de pathologieCHU Bordeaux‐GU PellegrinBordeauxFrance
| | - Aurélien Trimouille
- Unité fonctionnelle d'histologie moléculaire, Service de pathologieCHU Bordeaux‐GU PellegrinBordeauxFrance
| | - Gaëlle Hardy
- Laboratoire de Génétique Moléculaire: Maladies Héréditaires et OncologieInstitut de Biologie et de Pathologie, CHU Grenoble AlpesGrenobleFrance
| | - Stéphane Allouche
- Service de biochimieInstitut Territorial de Biologie en Santé, CHU Caen, Hôpital de la Côte de NacreCaenFrance
| | - Cécile Acquaviva‐Bourdain
- Service de biochimie et biologie moléculaire Grand Est, UM Maladies Héréditaires du Métabolisme, Centre de biologie et pathologie EstCHU Lyon HCL, GH EstLyonFrance
| | - Cécile Pagan
- Service de biochimie et biologie moléculaire Grand Est, UM Maladies Héréditaires du Métabolisme, Centre de biologie et pathologie EstCHU Lyon HCL, GH EstLyonFrance
| | - Anne‐Sophie Lebre
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266 [Krebs team]Université de Reims Champagne‐Ardenne (UFR médicale) ‐ CHU de Reims‐Université Paris CitéParisFrance
| | - Pascal Reynier
- Service de biochimie et biologie moléculaire, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| | - Mireille Cossee
- Laboratoire de Génétique Moléculaire, CHU Montpellier, PhyMedExpUniversité de Montpellier, INSERM, CNRSMontpellierFrance
| | - Shahram Attarian
- Service des Maladies Neuromusculaires et la SLA, FILNEMUS, Euro‐NMDAIX‐CHU La TimoneMarseille UniversitéMarseilleFrance
| | - Véronique Paquis‐Flucklinger
- Service de génétique médicale, Centre de référence des maladies mitochondriales, CHU NiceUniversité Côte d'Azur, CNRS, INSERM, IRCANNiceFrance
| | | | - Vincent Procaccio
- Service de génétique, Institut de Biologie en santé, CHU AngersUniv Angers, INSERM, CNRS, MITOVASC, Equipe MitoLab, SFR ICATAngersFrance
| |
Collapse
|
10
|
Sung AY, Guerra RM, Steenberge LH, Alston CL, Murayama K, Okazaki Y, Shimura M, Prokisch H, Ghezzi D, Torraco A, Carrozzo R, Rötig A, Taylor RW, Keck JL, Pagliarini DJ. Systematic analysis of NDUFAF6 in complex I assembly and mitochondrial disease. Nat Metab 2024; 6:1128-1142. [PMID: 38720117 PMCID: PMC11395703 DOI: 10.1038/s42255-024-01039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/28/2024] [Indexed: 06/27/2024]
Abstract
Isolated complex I (CI) deficiencies are a major cause of primary mitochondrial disease. A substantial proportion of CI deficiencies are believed to arise from defects in CI assembly factors (CIAFs) that are not part of the CI holoenzyme. The biochemistry of these CIAFs is poorly defined, making their role in CI assembly unclear, and confounding interpretation of potential disease-causing genetic variants. To address these challenges, we devised a deep mutational scanning approach to systematically assess the function of thousands of NDUFAF6 genetic variants. Guided by these data, biochemical analyses and cross-linking mass spectrometry, we discovered that the CIAF NDUFAF6 facilitates incorporation of NDUFS8 into CI and reveal that NDUFS8 overexpression rectifies NDUFAF6 deficiency. Our data further provide experimental support of pathogenicity for seven novel NDUFAF6 variants associated with human pathology and introduce functional evidence for over 5,000 additional variants. Overall, our work defines the molecular function of NDUFAF6 and provides a clinical resource for aiding diagnosis of NDUFAF6-related diseases.
Collapse
Affiliation(s)
- Andrew Y Sung
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rachel M Guerra
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Laura H Steenberge
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Charlotte L Alston
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Kei Murayama
- Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yasushi Okazaki
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Masaru Shimura
- Department of Metabolism, Chiba Children's Hospital, Chiba, Japan
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Holger Prokisch
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
- School of Medicine, Institute of Human Genetics, Technical University of Munich, Munich, Germany
| | - Daniele Ghezzi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Instituto Neurologico Carlo Besta, Milan, Italy
| | - Alessandra Torraco
- Unit of Cell Biology and Diagnosis of Mitochondrial Disorders, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rosalba Carrozzo
- Unit of Cell Biology and Diagnosis of Mitochondrial Disorders, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Agnès Rötig
- Université Paris Cité, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Robert W Taylor
- Mitochondrial Research Group, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - James L Keck
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - David J Pagliarini
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
11
|
Yang Y, del Gaudio D, Santani A, Scott SA. Applications of genome sequencing as a single platform for clinical constitutional genetic testing. GENETICS IN MEDICINE OPEN 2024; 2:101840. [PMID: 39822265 PMCID: PMC11736070 DOI: 10.1016/j.gimo.2024.101840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/02/2024] [Accepted: 03/11/2024] [Indexed: 01/19/2025]
Abstract
The number of human disease genes has dramatically increased over the past decade, largely fueled by ongoing advances in sequencing technologies. In parallel, the number of available clinical genetic tests has also increased, including the utilization of exome sequencing for undiagnosed diseases. Although most clinical sequencing tests have been centered on enrichment-based multigene panels and exome sequencing, the continued improvements in performance and throughput of genome sequencing suggest that this technology is emerging as a potential platform for routine clinical genetic testing. A notable advantage is a single workflow with the opportunity to reflexively interrogate content as clinically indicated; however, challenges with implementing routine clinical genome sequencing still remain. This review is centered on evaluating the applications of genome sequencing as a single platform for clinical constitutional genetic testing, including its potential utility for diagnostic testing, carrier screening, cytogenomic molecular karyotyping, prenatal testing, mitochondrial genome interrogation, and pharmacogenomic and polygenic risk score testing.
Collapse
Affiliation(s)
- Yao Yang
- Department of Pathology, Stanford University, Stanford, CA
- Clinical Genomics Laboratory, Stanford Medicine, Palo Alto, CA
| | | | | | - Stuart A. Scott
- Department of Pathology, Stanford University, Stanford, CA
- Clinical Genomics Laboratory, Stanford Medicine, Palo Alto, CA
| |
Collapse
|
12
|
Missen S, Wilson C, Potter H, Vincent AL, Murphy R, Roxburgh R, Rodrigues M, Poke G, Robertson SP, Thorburn DR, Glamuzina E. Mitochondrial disease in New Zealand: a nationwide prevalence study. Intern Med J 2024; 54:388-397. [PMID: 37732891 DOI: 10.1111/imj.16211] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/01/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND The complexities of mitochondrial disease make epidemiological studies challenging, yet this information is important in understanding the healthcare burden and addressing service and educational needs. Existing studies are limited to quaternary centres or focus on a single genotype or phenotype and estimate disease prevalence at 12.5 per 100 000. New Zealand's (NZ) size and partially integrated national healthcare system make it amenable to a nationwide prevalence study. AIM To estimate the prevalence of molecularly confirmed and suspected mitochondrial disease on 31 December 2015 in NZ. METHODS Cases were identified from subspecialists and laboratory databases and through interrogation of the Ministry of Health National Minimum Dataset with a focus on presentations between 2000 and 2015. Patient records were reviewed, and those with a diagnosis of 'mitochondrial disease' who were alive and residing in NZ on the prevalence date were included. These were divided into molecularly confirmed and clinically suspected cases. Official NZ estimated resident population data were used to calculate prevalence. RESULTS Seven hundred twenty-three unique national health index numbers were identified. Five hundred five were excluded. The minimum combined prevalence for mitochondrial disease was 4.7 per 100 000 (95% confidence interval (CI): 4.1-5.4). The minimum prevalence for molecularly confirmed and suspected disease was 2.9 (95% CI 2.4-3.4) and 1.8 (95% CI 1.4-2.2) cases per 100 000 respectively. CONCLUSIONS Within the limitations of this study, comparison to similar prevalence studies performed by specialist referral centres suggests mitochondrial disease is underdiagnosed in NZ. This highlights a need for improved education and referral pathways for mitochondrial disease in NZ.
Collapse
Affiliation(s)
- Sarah Missen
- Child Health Service, Whangarei Hospital, Te Whatu Ora - Health New Zealand, Te Tai Tokerau, Whangarei, New Zealand
- Department of Paediatrics: Child and Youth Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Callum Wilson
- Department of Paediatrics: Child and Youth Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Adult and Paediatric National Metabolic Service, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
| | - Howard Potter
- Canterbury Health Labs, Te Whatu Ora - Health New Zealand, Waitaha Canterbury, Christchurch, New Zealand
| | - Andrea L Vincent
- Greenlane Eye Clinic, Greenlane Clinical Centre, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Rinki Murphy
- Auckland Diabetes Centre, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Te Mana Ki Tua, Specialist Weight Management Service, Middlemore Hospital, Te Whatu Ora - Health New Zealand, Counties Manukau, Auckland, New Zealand
| | - Richard Roxburgh
- Department of Neurology, Auckland City Hospital, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
- Centre for Brain Research, Neurogenetics Clinics, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Miriam Rodrigues
- Department of Neurology, Auckland City Hospital, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
- Centre for Brain Research, Neurogenetics Clinics, Faculty of Medicine and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Gemma Poke
- Central Hub, Genetic Health Service New Zealand, Te Whatu Ora - Health New Zealand, Wellington, New Zealand
| | | | - David R Thorburn
- Genomic Medicine Theme, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Emma Glamuzina
- Department of Paediatrics: Child and Youth Health, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Adult and Paediatric National Metabolic Service, Te Whatu Ora - Health New Zealand, Te Toka Tumai, Auckland, New Zealand
| |
Collapse
|
13
|
Lin Z, Liu L, Li X, Huang S, Zhao H, Zeng S, Yang H, Xie Y, Zhang R. Phenotype-driven reanalysis reveals five novel pathogenic variants in 40 exome-negative families with Charcot-Marie-Tooth Disease. J Neurol 2024; 271:497-503. [PMID: 37776383 DOI: 10.1007/s00415-023-11991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/01/2023] [Accepted: 09/06/2023] [Indexed: 10/02/2023]
Abstract
BACKGROUND To identify genetic causes in 40 whole exome sequencing (WES)-negative Charcot-Marie-Tooth (CMT) families and provide a summary of the clinical and genetic features of the diagnosed patients. METHODS The clinical information and sequencing data of 40 WES-negative families out of 131 CMT families were collected, and phenotype-driven reanalysis was conducted using the Exomiser software. RESULTS The molecular diagnosis was regained in 4 families, increasing the overall diagnosis rate by 3.0%. One family with adolescent-onset pure CMT1 was diagnosed [POLR3B: c.2810G>A (p.R937Q)] due to the novel genotype-phenotype association. One infantile-onset, severe CMT1 family with deep sensory disturbance was diagnosed by screening the BAM file and harbored c.1174C>T (p.R392*) and 875_927delinsCTGCCCACTCTGCCCACTCTGCCCACTCTG (p.V292Afs53) of PRX. Two families were diagnosed due to characteristic phenotypes, including an infantile-onset ICMT family with renal dysfunction harboring c.213_233delinsGAGGAGCA (p.S72Rfs34) of INF2 and an adolescent-onset CMT2 family with optic atrophy harboring c.560C>T (p.P187L) and c.616A>G (p.K206E) of SLC25A46. According to the American College of Medical Genetics and Genomics (ACMG) guidelines, the variants of POLR3B and SLC25A46 were classified as likely pathogenic, and the variants of INF2 and PRX were pathogenic. All these variants were first reported worldwide except for p.R392* of PRX. CONCLUSIONS We identified five novel pathogenic variants in POLR3B, PRX, INF2, and SLC25A46, which broaden their phenotypic and genotypic spectrums. Regular phenotype-driven reanalysis is a powerful strategy for increasing the diagnostic yield of WES-negative CMT patients, and long-term follow-up and screening BAM files for contiguous deletion and missense variants are both essential for reanalysis.
Collapse
Affiliation(s)
- Zhiqiang Lin
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Lei Liu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Health Management Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaobo Li
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Shunxiang Huang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Huadong Zhao
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Sen Zeng
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Honglan Yang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Yongzhi Xie
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
14
|
Baker ZN, Forny P, Pagliarini DJ. Mitochondrial proteome research: the road ahead. Nat Rev Mol Cell Biol 2024; 25:65-82. [PMID: 37773518 PMCID: PMC11378943 DOI: 10.1038/s41580-023-00650-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 10/01/2023]
Abstract
Mitochondria are multifaceted organelles with key roles in anabolic and catabolic metabolism, bioenergetics, cellular signalling and nutrient sensing, and programmed cell death processes. Their diverse functions are enabled by a sophisticated set of protein components encoded by the nuclear and mitochondrial genomes. The extent and complexity of the mitochondrial proteome remained unclear for decades. This began to change 20 years ago when, driven by the emergence of mass spectrometry-based proteomics, the first draft mitochondrial proteomes were established. In the ensuing decades, further technological and computational advances helped to refine these 'maps', with current estimates of the core mammalian mitochondrial proteome ranging from 1,000 to 1,500 proteins. The creation of these compendia provided a systemic view of an organelle previously studied primarily in a reductionist fashion and has accelerated both basic scientific discovery and the diagnosis and treatment of human disease. Yet numerous challenges remain in understanding mitochondrial biology and translating this knowledge into the medical context. In this Roadmap, we propose a path forward for refining the mitochondrial protein map to enhance its discovery and therapeutic potential. We discuss how emerging technologies can assist the detection of new mitochondrial proteins, reveal their patterns of expression across diverse tissues and cell types, and provide key information on proteoforms. We highlight the power of an enhanced map for systematically defining the functions of its members. Finally, we examine the utility of an expanded, functionally annotated mitochondrial proteome in a translational setting for aiding both diagnosis of mitochondrial disease and targeting of mitochondria for treatment.
Collapse
Affiliation(s)
- Zakery N Baker
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Patrick Forny
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David J Pagliarini
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
15
|
Olimpio C, Paramonov I, Matalonga L, Laurie S, Schon K, Polavarapu K, Kirschner J, Schara-Schmidt U, Lochmüller H, Chinnery PF, Horvath R. Increased Diagnostic Yield by Reanalysis of Whole Exome Sequencing Data in Mitochondrial Disease. J Neuromuscul Dis 2024; 11:767-775. [PMID: 38759022 PMCID: PMC11307028 DOI: 10.3233/jnd-240020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2024] [Indexed: 05/19/2024]
Abstract
Background The genetic diagnosis of mitochondrial disorders is complicated by its genetic and phenotypic complexity. Next generation sequencing techniques have much improved the diagnostic yield for these conditions. A cohort of individuals with multiple respiratory chain deficiencies, reported in the literature 10 years ago, had a diagnostic rate of 60% by whole exome sequencing (WES) but 40% remained undiagnosed. Objective We aimed to identify a genetic diagnosis by reanalysis of the WES data for the undiagnosed arm of this 10-year-old cohort of patients with suspected mitochondrial disorders. Methods The WES data was transferred and processed by the RD-Connect Genome-Phenome Analysis Platform (GPAP) using their standardized pipeline. Variant prioritisation was carried out on the RD-Connect GPAP. Results Singleton WES data from 14 individuals was reanalysed. We identified a possible or likely genetic diagnosis in 8 patients (8/14, 57%). The variants identified were in a combination of mitochondrial DNA (n = 1, MT-TN), nuclear encoded mitochondrial genes (n = 2, PDHA1, and SUCLA2) and nuclear genes associated with nonmitochondrial disorders (n = 5, PNPLA2, CDC40, NBAS and SLC7A7). Variants in both the NBAS and CDC40 genes were established as disease causing after the original cohort was published. We increased the diagnostic yield for the original cohort by 15% without generating any further genomic data. Conclusions In the era of multiomics we highlight that reanalysis of existing WES data is a valid tool for generating additional diagnosis in patients with suspected mitochondrial disease, particularly when more time has passed to allow for new bioinformatic pipelines to emerge, for the development of new tools in variant interpretation aiding in reclassification of variants and the expansion of scientific knowledge on additional genes.
Collapse
Affiliation(s)
- Catarina Olimpio
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ida Paramonov
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | | | - Steven Laurie
- Centro Nacional de Análisis Genómico, Barcelona, Spain
| | - Katherine Schon
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- East Anglian Medical Genetics Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kiran Polavarapu
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Center for Neuromuscular Disorders, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, Essen, Germany
| | - Hanns Lochmüller
- Centro Nacional de Análisis Genómico, Barcelona, Spain
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center-University of Freiburg, Freiburg, Germany
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
| | - Patrick F. Chinnery
- MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rita Horvath
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| |
Collapse
|
16
|
Major TC, Arany ES, Schon K, Simo M, Karcagi V, van den Ameele J, Yu Wai Man P, Chinnery PF, Olimpio C, Horvath R. Case report: Mutations in DNAJC30 causing autosomal recessive Leber hereditary optic neuropathy are common amongst Eastern European individuals. Front Neurol 2023; 14:1292320. [PMID: 38107630 PMCID: PMC10722306 DOI: 10.3389/fneur.2023.1292320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/03/2023] [Indexed: 12/19/2023] Open
Abstract
Background Leber Hereditary Optic Neuropathy (LHON) is the most common inherited mitochondrial disease characterized by bilateral, painless, subacute visual loss with a peak age of onset in the second to third decade. Historically, LHON was thought to be exclusively maternally inherited due to mutations in mitochondrial DNA (mtDNA); however, recent studies have identified an autosomal recessive form of LHON (arLHON) caused by point mutations in the nuclear gene, DNAJC30. Case Presentations In this study, we report the cases of three Eastern European individuals presenting with bilateral painless visual loss, one of whom was also exhibiting motor symptoms. After a several-year-long diagnostic journey, all three patients were found to carry the homozygous c.152A>G (p.Tyr51Cys) mutation in DNAJC30. This has been identified as the most common arLHON pathogenic variant and has been shown to exhibit a significant founder effect amongst Eastern European individuals. Conclusion This finding adds to the growing cohort of patients with arLHON and demonstrates the importance of DNAJC30 screening in patients with molecularly undiagnosed LHON, particularly in Eastern European individuals. It is of heightened translational significance as patients diagnosed with arLHON exhibit a better prognosis and response to therapeutic treatment with the co-enzyme Q10 analog idebenone.
Collapse
Affiliation(s)
- Toby Charles Major
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Eszter Sara Arany
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Katherine Schon
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Genetics, East Anglian Medical Genetics Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Magdolna Simo
- University Clinic of Neurology, Semmelweis University, Budapest, Hungary
| | | | - Jelle van den Ameele
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Patrick Yu Wai Man
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- NIHR Biomedical Research Centre, Moorfields Eye Hospital & UCL Institute of Ophthalmology, London, United Kingdom
- Cambridge Eye Unit, Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Patrick F. Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Catarina Olimpio
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Genetics, East Anglian Medical Genetics Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Rita Horvath
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
17
|
Serag M, Plutino M, Charles P, Azulay JP, Chaussenot A, Paquis-Flucklinger V, Ait-El-Mkadem Saadi S, Rouzier C. A Case Report of SYNE1 Deficiency-Mimicking Mitochondrial Disease and the Value of Pangenomic Investigations. Genes (Basel) 2023; 14:2154. [PMID: 38136976 PMCID: PMC10743207 DOI: 10.3390/genes14122154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Mitochondrial disorders are characterized by a huge clinical, biochemical, and genetic heterogeneity, which poses significant diagnostic challenges. Several studies report that more than 50% of patients with suspected mitochondrial disease could have a non-mitochondrial disorder. Thus, only the identification of the causative pathogenic variant can confirm the diagnosis. Herein, we describe the diagnostic journey of a family suspected of having a mitochondrial disorder who were referred to our Genetics Department. The proband presented with the association of cerebellar ataxia, COX-negative fibers on muscle histology, and mtDNA deletions. Whole exome sequencing (WES), supplemented by a high-resolution array, comparative genomic hybridization (array-CGH), allowed us to identify two pathogenic variants in the non-mitochondrial SYNE1 gene. The proband and her affected sister were found to be compound heterozygous for a known nonsense variant (c.13258C>T, p.(Arg4420Ter)), and a large intragenic deletion that was predicted to result in a loss of function. To our knowledge, this is the first report of a large intragenic deletion of SYNE1 in patients with cerebellar ataxia (ARCA1). This report highlights the interest in a pangenomic approach to identify the genetic basis in heterogeneous neuromuscular patients with the possible cause of mitochondrial disease. Moreover, even rare copy number variations should be considered in patients with a phenotype suggestive of SYNE1 deficiency.
Collapse
Affiliation(s)
- Mounir Serag
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| | - Morgane Plutino
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| | - Perrine Charles
- Service de Génétique, La Pitié-Salpêtrière, AP-HP, 75610 Paris, France;
| | | | - Annabelle Chaussenot
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| | - Véronique Paquis-Flucklinger
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| | - Samira Ait-El-Mkadem Saadi
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| | - Cécile Rouzier
- Service de Génétique Médicale, Hôpital l’Archet 2, CHU de Nice, 151 Route Saint-Antoine de Ginestière, 06202 Nice, France; (M.S.); (M.P.); (A.C.); (V.P.-F.); (S.A.-E.-M.S.)
- CNRS UMR7284/ INSERM U1081, Faculté de Médecine, Université Côte d’Azur, 06107 Nice, France
| |
Collapse
|
18
|
Jain V, Irving R, Williams A. Genomic testing in neurology. Pract Neurol 2023; 23:420-429. [PMID: 37468300 DOI: 10.1136/pn-2023-003735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2023] [Indexed: 07/21/2023]
Abstract
Genomic testing has been available for neurological conditions for decades. However, in recent years, there has been a significant change in its availability, range and cost, as well as improvements in the technology and knowledge that underpin how the genome is interrogated. Neurologists can encounter a wide range of genetic conditions, and so their understanding of genomic testing is fundamental to modern clinical practice.
Collapse
Affiliation(s)
- Vani Jain
- All Wales Medical Genomics Service, University Hospital of Wales Healthcare NHS Trust, Cardiff, UK
| | - Rachel Irving
- All Wales Medical Genomics Service, University Hospital of Wales Healthcare NHS Trust, Cardiff, UK
| | - Angharad Williams
- All Wales Medical Genomics Service, University Hospital of Wales Healthcare NHS Trust, Cardiff, UK
| |
Collapse
|
19
|
Balciuniene J, Liu R, Bean L, Guo F, Nallamilli BRR, Guruju N, Chen-Deutsch X, Yousaf R, Fura K, Chin E, Mathur A, Ma Z, Carmichael J, da Silva C, Collins C, Hegde M. At-Risk Genomic Findings for Pediatric-Onset Disorders From Genome Sequencing vs Medically Actionable Gene Panel in Proactive Screening of Newborns and Children. JAMA Netw Open 2023; 6:e2326445. [PMID: 37523181 PMCID: PMC10391308 DOI: 10.1001/jamanetworkopen.2023.26445] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
Importance Although the clinical utility of genome sequencing for critically ill children is well recognized, its utility for proactive pediatric screening is not well explored. Objective To evaluate molecular findings from screening ostensibly healthy children with genome sequencing compared with a gene panel for medically actionable pediatric conditions. Design, Setting, and Participants This case series study was conducted among consecutive, apparently healthy children undergoing proactive genetic screening for pediatric disorders by genome sequencing (n = 562) or an exome-based panel of 268 genes (n = 606) from March 1, 2018, through July 31, 2022. Exposures Genetic screening for pediatric-onset disorders using genome sequencing or an exome-based panel of 268 genes. Main Outcomes and Measures Molecular findings indicative of genetic disease risk. Results Of 562 apparently healthy children (286 girls [50.9%]; median age, 29 days [IQR, 9-117 days]) undergoing screening by genome sequencing, 46 (8.2%; 95% CI, 5.9%-10.5%) were found to be at risk for pediatric-onset disease, including 22 children (3.9%) at risk for high-penetrance disorders. Sequence analysis uncovered molecular diagnoses among 32 individuals (5.7%), while copy number variant analysis uncovered molecular diagnoses among 14 individuals (2.5%), including 4 individuals (0.7%) with chromosome scale abnormalities. Overall, there were 47 molecular diagnoses, with 1 individual receiving 2 diagnoses; of the 47 potential diagnoses, 22 (46.8%) were associated with high-penetrance conditions. Pathogenic variants in medically actionable pediatric genes were found in 6 individuals (1.1%), constituting 12.8% (6 of 47) of all diagnoses. At least 1 pharmacogenomic variant was reported for 89.0% (500 of 562) of the cohort. In contrast, of 606 children (293 girls [48.3%]; median age, 26 days [IQR, 10-67 days]) undergoing gene panel screening, only 13 (2.1%; 95% CI, 1.0%-3.3%) resulted in potential childhood-onset diagnoses, a significantly lower rate than those screened by genome sequencing (P < .001). Conclusions and Relevance In this case series study, genome sequencing as a proactive screening approach for children, due to its unrestrictive gene content and technical advantages in comparison with an exome-based gene panel for medically actionable childhood conditions, uncovered a wide range of heterogeneous high-penetrance pediatric conditions that could guide early interventions and medical management.
Collapse
Affiliation(s)
| | - Ruby Liu
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Lora Bean
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Fen Guo
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | | | - Naga Guruju
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | | | - Rizwan Yousaf
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Kristina Fura
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Ephrem Chin
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Abhinav Mathur
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | - Zeqiang Ma
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| | | | | | | | - Madhuri Hegde
- PerkinElmer Genomics, PerkinElmer Inc, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Macken WL, Falabella M, Pizzamiglio C, Woodward CE, Scotchman E, Chitty LS, Polke JM, Bugiardini E, Hanna MG, Vandrovcova J, Chandler N, Labrum R, Pitceathly RDS. Enhanced mitochondrial genome analysis: bioinformatic and long-read sequencing advances and their diagnostic implications. Expert Rev Mol Diagn 2023; 23:797-814. [PMID: 37642407 DOI: 10.1080/14737159.2023.2241365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION Primary mitochondrial diseases (PMDs) comprise a large and heterogeneous group of genetic diseases that result from pathogenic variants in either nuclear DNA (nDNA) or mitochondrial DNA (mtDNA). Widespread adoption of next-generation sequencing (NGS) has improved the efficiency and accuracy of mtDNA diagnoses; however, several challenges remain. AREAS COVERED In this review, we briefly summarize the current state of the art in molecular diagnostics for mtDNA and consider the implications of improved whole genome sequencing (WGS), bioinformatic techniques, and the adoption of long-read sequencing, for PMD diagnostics. EXPERT OPINION We anticipate that the application of PCR-free WGS from blood DNA will increase in diagnostic laboratories, while for adults with myopathic presentations, WGS from muscle DNA may become more widespread. Improved bioinformatic strategies will enhance WGS data interrogation, with more accurate delineation of mtDNA and NUMTs (nuclear mitochondrial DNA segments) in WGS data, superior coverage uniformity, indirect measurement of mtDNA copy number, and more accurate interpretation of heteroplasmic large-scale rearrangements (LSRs). Separately, the adoption of diagnostic long-read sequencing could offer greater resolution of complex LSRs and the opportunity to phase heteroplasmic variants.
Collapse
Affiliation(s)
- William L Macken
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Chiara Pizzamiglio
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Cathy E Woodward
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Elizabeth Scotchman
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Lyn S Chitty
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - James M Polke
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Enrico Bugiardini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Michael G Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Natalie Chandler
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Robyn Labrum
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
21
|
Henry OJ, Stödberg T, Båtelson S, Rasi C, Stranneheim H, Wedell A. Individualised human phenotype ontology gene panels improve clinical whole exome and genome sequencing analytical efficacy in a cohort of developmental and epileptic encephalopathies. Mol Genet Genomic Med 2023; 11:e2167. [PMID: 36967109 PMCID: PMC10337286 DOI: 10.1002/mgg3.2167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/21/2023] [Accepted: 03/01/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND The majority of genetic epilepsies remain unsolved in terms of specific genotype. Phenotype-based genomic analyses have shown potential to strengthen genomic analysis in various ways, including improving analytical efficacy. METHODS We have tested a standardised phenotyping method termed 'Phenomodels' for integrating deep-phenotyping information with our in-house developed clinical whole exome/genome sequencing analytical pipeline. Phenomodels includes a user-friendly epilepsy phenotyping template and an objective measure for selecting which template terms to include in individualised Human Phenotype Ontology (HPO) gene panels. In a pilot study of 38 previously solved cases of developmental and epileptic encephalopathies, we compared the sensitivity and specificity of the individualised HPO gene panels with the clinical epilepsy gene panel. RESULTS The Phenomodels template showed high sensitivity for capturing relevant phenotypic information, where 37/38 individuals' HPO gene panels included the causative gene. The HPO gene panels also had far fewer variants to assess than the epilepsy gene panel. CONCLUSION We have demonstrated a viable approach for incorporating standardised phenotype information into clinical genomic analyses, which may enable more efficient analysis.
Collapse
Affiliation(s)
- Olivia J. Henry
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | - Tommy Stödberg
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Department of Pediatric NeurologyKarolinska University HospitalStockholmSweden
| | - Sofia Båtelson
- Department of Pediatric NeurologyKarolinska University HospitalStockholmSweden
| | - Chiara Rasi
- Science for Life Laboratory, Department of Microbiology, Tumour and Cell BiologyKarolinska InstitutetStockholmSweden
| | - Henrik Stranneheim
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Science for Life Laboratory, Department of Microbiology, Tumour and Cell BiologyKarolinska InstitutetStockholmSweden
- Centre for Inherited Metabolic DiseasesKarolinska University HospitalStockholmSweden
| | - Anna Wedell
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Centre for Inherited Metabolic DiseasesKarolinska University HospitalStockholmSweden
| |
Collapse
|
22
|
Viscomi C, van den Ameele J, Meyer KC, Chinnery PF. Opportunities for mitochondrial disease gene therapy. Nat Rev Drug Discov 2023; 22:429-430. [PMID: 37106085 DOI: 10.1038/d41573-023-00067-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
|
23
|
Squires JE, Miethke AG, Valencia CA, Hawthorne K, Henn L, Van Hove JL, Squires RH, Bove K, Horslen S, Kohli R, Molleston JP, Romero R, Alonso EM, Bezerra JA, Guthery SL, Hsu E, Karpen SJ, Loomes KM, Ng VL, Rosenthal P, Mysore K, Wang KS, Friederich MW, Magee JC, Sokol RJ. Clinical spectrum and genetic causes of mitochondrial hepatopathy phenotype in children. Hepatol Commun 2023; 7:e0139. [PMID: 37184518 PMCID: PMC10187840 DOI: 10.1097/hc9.0000000000000139] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/19/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND Alterations in both mitochondrial DNA (mtDNA) and nuclear DNA genes affect mitochondria function, causing a range of liver-based conditions termed mitochondrial hepatopathies (MH), which are subcategorized as mtDNA depletion, RNA translation, mtDNA deletion, and enzymatic disorders. We aim to enhance the understanding of pathogenesis and natural history of MH. METHODS We analyzed data from patients with MH phenotypes to identify genetic causes, characterize the spectrum of clinical presentation, and determine outcomes. RESULTS Three enrollment phenotypes, that is, acute liver failure (ALF, n = 37), chronic liver disease (Chronic, n = 40), and post-liver transplant (n = 9), were analyzed. Patients with ALF were younger [median 0.8 y (range, 0.0, 9.4) vs 3.4 y (0.2, 18.6), p < 0.001] with fewer neurodevelopmental delays (40.0% vs 81.3%, p < 0.001) versus Chronic. Comprehensive testing was performed more often in Chronic than ALF (90.0% vs 43.2%); however, etiology was identified more often in ALF (81.3% vs 61.1%) with mtDNA depletion being most common (ALF: 77% vs Chronic: 41%). Of the sequenced cohort (n = 60), 63% had an identified mitochondrial disorder. Cluster analysis identified a subset without an underlying genetic etiology, despite comprehensive testing. Liver transplant-free survival was 40% at 2 years (ALF vs Chronic, 16% vs 65%, p < 0.001). Eighteen (21%) underwent transplantation. With 33 patient-years of follow-up after the transplant, 3 deaths were reported. CONCLUSIONS Differences between ALF and Chronic MH phenotypes included age at diagnosis, systemic involvement, transplant-free survival, and genetic etiology, underscoring the need for ultra-rapid sequencing in the appropriate clinical setting. Cluster analysis revealed a group meeting enrollment criteria but without an identified genetic or enzymatic diagnosis, highlighting the need to identify other etiologies.
Collapse
Affiliation(s)
- James E. Squires
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - C. Alexander Valencia
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Interpath Laboratory, Pendleton, Oregon, USA
| | - Kieran Hawthorne
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Lisa Henn
- Arbor Research Collaborative for Health, Ann Arbor, Michigan, USA
| | - Johan L.K. Van Hove
- University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Robert H. Squires
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kevin Bove
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Simon Horslen
- UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rohit Kohli
- Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Jean P. Molleston
- Indiana University-Riley Hospital for Children, Indianapolis, Indiana, USA
| | - Rene Romero
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Estella M. Alonso
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Jorge A. Bezerra
- Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stephen L. Guthery
- University of Utah School of Medicine, Primary Children’s Hospital, Salt Lake City, Utah, USA
| | - Evelyn Hsu
- University of Washington School of Medicine and Seattle Children’s Hospital, Seattle, Washington, USA
| | - Saul J. Karpen
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Kathleen M. Loomes
- The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Vicky L. Ng
- Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | | - Krupa Mysore
- Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Kasper S. Wang
- Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - Marisa W. Friederich
- University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| | - John C. Magee
- University of Michigan Hospitals and Health Centers, Ann Arbor, Michigan, USA
| | - Ronald J. Sokol
- University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
| |
Collapse
|
24
|
Nurchis MC, Altamura G, Riccardi MT, Radio FC, Chillemi G, Bertini ES, Garlasco J, Tartaglia M, Dallapiccola B, Damiani G. Whole genome sequencing diagnostic yield for paediatric patients with suspected genetic disorders: systematic review, meta-analysis, and GRADE assessment. Arch Public Health 2023; 81:93. [PMID: 37231492 DOI: 10.1186/s13690-023-01112-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 05/18/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND About 80% of the roughly 7,000 known rare diseases are single gene disorders, about 85% of which are ultra-rare, affecting less than one in one million individuals. NGS technologies, in particular whole genome sequencing (WGS) in paediatric patients suffering from severe disorders of likely genetic origin improve the diagnostic yield allowing targeted, effective care and management. The aim of this study is to perform a systematic review and meta-analysis to assess the effectiveness of WGS, with respect to whole exome sequencing (WES) and/or usual care, for the diagnosis of suspected genetic disorders among the paediatric population. METHODS A systematic review of the literature was conducted querying relevant electronic databases, including MEDLINE, EMBASE, ISI Web of Science, and Scopus from January 2010 to June 2022. A random-effect meta-analysis was run to inspect the diagnostic yield of different techniques. A network meta-analysis was also performed to directly assess the comparison between WGS and WES. RESULTS Of the 4,927 initially retrieved articles, thirty-nine met the inclusion criteria. Overall results highlighted a significantly higher pooled diagnostic yield for WGS, 38.6% (95% CI: [32.6 - 45.0]), in respect to WES, 37.8% (95% CI: [32.9 - 42.9]) and usual care, 7.8% (95% CI: [4.4 - 13.2]). The meta-regression output suggested a higher diagnostic yield of the WGS compared to WES after controlling for the type of disease (monogenic vs non-monogenic), with a tendency to better diagnostic performances for Mendelian diseases. The network meta-analysis showed a higher diagnostic yield for WGS compared to WES (OR = 1.54, 95%CI: [1.11 - 2.12]). CONCLUSIONS Although whole genome sequencing for the paediatric population with suspected genetic disorders provided an accurate and early genetic diagnosis in a high proportion of cases, further research is needed for evaluating costs, effectiveness, and cost-effectiveness of WGS and achieving an informed decision-making process. TRIAL REGISTRATION This systematic review has not been registered.
Collapse
Grants
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
- RF-2018-12,366,391, 2018 Ministero della Salute
Collapse
Affiliation(s)
- Mario Cesare Nurchis
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- School of Economics, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Gerardo Altamura
- Department of Health Sciences and Public Health, Section of Hygiene, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy.
| | - Maria Teresa Riccardi
- Department of Health Sciences and Public Health, Section of Hygiene, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Francesca Clementina Radio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCCS, 00146, Rome, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100, Viterbo, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Centro Nazionale Delle Ricerche, 70126, Bari, Italy
| | - Enrico Silvio Bertini
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCCS, 00146, Rome, Italy
| | - Jacopo Garlasco
- Department of Public Health Sciences and Paediatrics, University of Turin, 10126, Turin, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCCS, 00146, Rome, Italy
| | - Bruno Dallapiccola
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù IRCCS, 00146, Rome, Italy
| | - Gianfranco Damiani
- Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
- Department of Health Sciences and Public Health, Section of Hygiene, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
25
|
Wu TH, Peng J, Yang L, Chen YH, Lu XL, Huang JT, You JY, Ou-Yang WX, Sun YY, Xue YN, Mao X, Yan HM, Ren RN, Xie J, Chen ZH, Zhang VW, Lyu GZ, He F. Use of dual genomic sequencing to screen mitochondrial diseases in pediatrics: a retrospective analysis. Sci Rep 2023; 13:4193. [PMID: 36918699 PMCID: PMC10015028 DOI: 10.1038/s41598-023-31134-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Mitochondrial diseases (MDs) were a large group multisystem disorders, attributable in part to the dual genomic control. The advent of massively sequencing has improved diagnostic rates and speed, and was increasingly being used as a first-line diagnostic test. Paediatric patients (aged < 18 years) who underwent dual genomic sequencing were enrolled in this retrospective multicentre study. We evaluated the mitochondrial disease criteria (MDC) and molecular diagnostic yield of dual genomic sequencing. Causative variants were identified in 177 out of 503 (35.2%) patients using dual genomic sequencing. Forty-six patients (9.1%) had mitochondria-related variants, including 25 patients with nuclear DNA (nDNA) variants, 15 with mitochondrial DNA (mtDNA) variants, and six with dual genomic variants (MT-ND6 and POLG; MT-ND5 and RARS2; MT-TL1 and NARS2; MT-CO2 and NDUFS1; MT-CYB and SMARCA2; and CHRNA4 and MT-CO3). Based on the MDC, 15.2% of the patients with mitochondria-related variants were classified as "unlikely to have mitochondrial disorder". Moreover, 4.5% of the patients with non-mitochondria-related variants and 1.43% with negative genetic tests, were classified as "probably having mitochondrial disorder". Dual genomic sequencing in suspected MDs provided a more comprehensive and accurate diagnosis for pediatric patients, especially for patients with dual genomic variants.
Collapse
Affiliation(s)
- Teng-Hui Wu
- Department of Pediatrics, Xiangya Hospital Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Li Yang
- Department of Pediatrics, Xiangya Hospital Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yan-Hui Chen
- Department of Pediatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, China
| | - Xiu-Lan Lu
- Department of Pediatric Intensive Care Unit, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan, China
| | - Jiao-Tian Huang
- Department of Pediatric Intensive Care Unit, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan, China
| | - Jie-Yu You
- Department of Gastroenterology and Nutrition, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan, China
| | - Wen-Xian Ou-Yang
- Department of Hepatopathy, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan, China
| | - Yue-Yu Sun
- Department of Pediatric Intensive Care Unit, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences (GAMS), 106 Zhongshan 2nd Road, Guangzhou, Guangdong, China
| | - Yi-Nan Xue
- Department of Pediatrics, Brain Hospital of Hunan Province, 427 Furong Road, Changsha, Hunan, China
| | - Xiao Mao
- Department of Medical Genetics, Maternal,, Child Health Hospital of Hunan Province, 53 Xiangchun Road, Changsha, Hunan, China
| | - Hui-Ming Yan
- Department of Medical Genetics, Maternal,, Child Health Hospital of Hunan Province, 53 Xiangchun Road, Changsha, Hunan, China
| | - Rong-Na Ren
- Department of Pediatrics, The 900Th Hospital of Joint Logistic Support Force, PLA, Fuzhou, Fujian, China
| | - Jing Xie
- Department of Pediatrics, The First Hospital of Hunan University of Chinese Medicine, 95 Shaoshan Road, Changsha, Hunan, China
| | - Zhi-Heng Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan, China
| | - Victor-Wei Zhang
- Amcare Genomics Laboratory, Guangzhou, Guangdong, China.,Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Gui-Zhen Lyu
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Fang He
- Department of Pediatrics, Xiangya Hospital Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
26
|
Mavraki E, Labrum R, Sergeant K, Alston CL, Woodward C, Smith C, Knowles CVY, Patel Y, Hodsdon P, Baines JP, Blakely EL, Polke J, Taylor RW, Fratter C. Genetic testing for mitochondrial disease: the United Kingdom best practice guidelines. Eur J Hum Genet 2023; 31:148-163. [PMID: 36513735 PMCID: PMC9905091 DOI: 10.1038/s41431-022-01249-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
Primary mitochondrial disease describes a diverse group of neuro-metabolic disorders characterised by impaired oxidative phosphorylation. Diagnosis is challenging; >350 genes, both nuclear and mitochondrial DNA (mtDNA) encoded, are known to cause mitochondrial disease, leading to all possible inheritance patterns and further complicated by heteroplasmy of the multicopy mitochondrial genome. Technological advances, particularly next-generation sequencing, have driven a shift in diagnostic practice from 'biopsy first' to genome-wide analyses of blood and/or urine DNA. This has led to the need for a reference framework for laboratories involved in mitochondrial genetic testing to facilitate a consistent high-quality service. In the United Kingdom, consensus guidelines have been prepared by a working group of Clinical Scientists from the NHS Highly Specialised Service followed by national laboratory consultation. These guidelines summarise current recommended technologies and methodologies for the analysis of mtDNA and nuclear-encoded genes in patients with suspected mitochondrial disease. Genetic testing strategies for diagnosis, family testing and reproductive options including prenatal diagnosis are outlined. Importantly, recommendations for the minimum levels of mtDNA testing for the most common referral reasons are included, as well as guidance on appropriate referrals and information on the minimal appropriate gene content of panels when analysing nuclear mitochondrial genes. Finally, variant interpretation and recommendations for reporting of results are discussed, focussing particularly on the challenges of interpreting and reporting mtDNA variants.
Collapse
Affiliation(s)
- Eleni Mavraki
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Robyn Labrum
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Kate Sergeant
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Charlotte L Alston
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Cathy Woodward
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Conrad Smith
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Charlotte V Y Knowles
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Yogen Patel
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Philip Hodsdon
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jack P Baines
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Emma L Blakely
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - James Polke
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | - Robert W Taylor
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Carl Fratter
- Oxford Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
27
|
Schon KR, Chinnery PF. Whole-genome sequencing for mitochondrial disorders identifies unexpected mimics. Pract Neurol 2023; 23:2-3. [PMID: 36253087 DOI: 10.1136/pn-2022-003570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 02/02/2023]
Affiliation(s)
- Katherine R Schon
- Department of Clinical Neurosciences, Cambridge Biomedical Campus, Cambridge, UK
- Academic Department of Medical Genetics, Cambridge Biomedical Campus, Cambridge, UK
| | - Patrick F Chinnery
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Paredes-Fuentes AJ, Oliva C, Urreizti R, Yubero D, Artuch R. Laboratory testing for mitochondrial diseases: biomarkers for diagnosis and follow-up. Crit Rev Clin Lab Sci 2023; 60:270-289. [PMID: 36694353 DOI: 10.1080/10408363.2023.2166013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The currently available biomarkers generally lack the specificity and sensitivity needed for the diagnosis and follow-up of patients with mitochondrial diseases (MDs). In this group of rare genetic disorders (mutations in approximately 350 genes associated with MDs), all clinical presentations, ages of disease onset and inheritance types are possible. Blood, urine, and cerebrospinal fluid surrogates are well-established biomarkers that are used in clinical practice to assess MD. One of the main challenges is validating specific and sensitive biomarkers for the diagnosis of disease and prediction of disease progression. Profiling of lactate, amino acids, organic acids, and acylcarnitine species is routinely conducted to assess MD patients. New biomarkers, including some proteins and circulating cell-free mitochondrial DNA, with increased diagnostic specificity have been identified in the last decade and have been proposed as potentially useful in the assessment of clinical outcomes. Despite these advances, even these new biomarkers are not sufficiently specific and sensitive to assess MD progression, and new biomarkers that indicate MD progression are urgently needed to monitor the success of novel therapeutic strategies. In this report, we review the mitochondrial biomarkers that are currently analyzed in clinical laboratories, new biomarkers, an overview of the most common laboratory diagnostic techniques, and future directions regarding targeted versus untargeted metabolomic and genomic approaches in the clinical laboratory setting. Brief descriptions of the current methodologies are also provided.
Collapse
Affiliation(s)
- Abraham J Paredes-Fuentes
- Division of Inborn Errors of Metabolism-IBC, Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Clara Oliva
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Roser Urreizti
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Delia Yubero
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Department of Genetic and Molecular Medicine-IPER, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Rafael Artuch
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Khamis S, Mitakidou MR, Champion M, Goyal S, Jones RL, Siddiqui A, Sabanathan S, Hedderly T, Lin JP, Jungbluth H, Papandreou A. Clinical Reasoning: A Teenage Girl With Progressive Hyperkinetic Movements, Seizures, and Encephalopathy. Neurology 2023; 100:30-37. [PMID: 36130841 PMCID: PMC9827126 DOI: 10.1212/wnl.0000000000201385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023] Open
Abstract
The "epilepsy-dyskinesia" spectrum is increasingly recognized in neurogenetic and neurometabolic conditions. It can be challenging to diagnose because of clinical and genetic heterogeneity, atypical or nonspecific presentations, and the rarity of each diagnostic entity. This is further complicated by the lack of sensitive or specific biomarkers for most nonenzymatic neurometabolic conditions. Nevertheless, clinical awareness and timely diagnosis are paramount to facilitate appropriate prognostication, counseling, and management.This report describes a case of a teenage girl who had presented at 14 months with a protracted illness manifesting as gastrointestinal upset and associated motor and cognitive regression. A choreoathetoid movement disorder, truncal ataxia, and microcephaly evolved after the acute phase. Neurometabolic and inflammatory investigations, EEG, brain MRI, muscle biopsy (including respiratory chain enzyme studies), and targeted genetic testing were unremarkable. A second distinct regression phase ensued at 14 years consisting of encephalopathy, multifocal motor seizures, absent deep tendon reflexes and worsening movements, gut dysmotility, and dysphagia. Video EEGs showed an evolving developmental and epileptic encephalopathy with multifocal seizures and nonepileptic movements. MRI of the brain revealed evolving and fluctuating patchy bihemispheric cortical changes, cerebellar atrophy with signal change, mild generalized brain volume loss, and abnormal lactate on MR spectroscopy. The article discusses the differential diagnostic approach and management options for patients presenting with neurologic regression, encephalopathy, seizures, and hyperkinetic movements. It also emphasizes the utility of next-generation sequencing in providing a rapid, efficient, cost-effective way of determining the underlying etiology of complex neurologic presentations.
Collapse
Affiliation(s)
- Sonia Khamis
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Maria R Mitakidou
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Michael Champion
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sushma Goyal
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rachel L Jones
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Ata Siddiqui
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Saraswathy Sabanathan
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Tammy Hedderly
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Jean-Pierre Lin
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Heinz Jungbluth
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Apostolos Papandreou
- From the Paediatric Neurology Department, Evelina London Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK; Metabolic Medicine Department, Evelina London Children's Hospital, London, UK; Clinical Neurophysiology Department, Evelina London Children's Hospital, London, UK; Clinical Genetics Department, Guys and St Thomas Hospital, London, UK; Neuroradiology Department, Evelina London Children's Hospital, London, UK; Women and Children's Health Institute, Faculty of Life Sciences & Medicine, King's College London, UK; Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, UK; and Molecular Neurosciences, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
30
|
Forny P, Bonilla X, Lamparter D, Shao W, Plessl T, Frei C, Bingisser A, Goetze S, van Drogen A, Harshman K, Pedrioli PGA, Howald C, Poms M, Traversi F, Bürer C, Cherkaoui S, Morscher RJ, Simmons L, Forny M, Xenarios I, Aebersold R, Zamboni N, Rätsch G, Dermitzakis ET, Wollscheid B, Baumgartner MR, Froese DS. Integrated multi-omics reveals anaplerotic rewiring in methylmalonyl-CoA mutase deficiency. Nat Metab 2023; 5:80-95. [PMID: 36717752 PMCID: PMC9886552 DOI: 10.1038/s42255-022-00720-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 12/01/2022] [Indexed: 01/31/2023]
Abstract
Methylmalonic aciduria (MMA) is an inborn error of metabolism with multiple monogenic causes and a poorly understood pathogenesis, leading to the absence of effective causal treatments. Here we employ multi-layered omics profiling combined with biochemical and clinical features of individuals with MMA to reveal a molecular diagnosis for 177 out of 210 (84%) cases, the majority (148) of whom display pathogenic variants in methylmalonyl-CoA mutase (MMUT). Stratification of these data layers by disease severity shows dysregulation of the tricarboxylic acid cycle and its replenishment (anaplerosis) by glutamine. The relevance of these disturbances is evidenced by multi-organ metabolomics of a hemizygous Mmut mouse model as well as through identification of physical interactions between MMUT and glutamine anaplerotic enzymes. Using stable-isotope tracing, we find that treatment with dimethyl-oxoglutarate restores deficient tricarboxylic acid cycling. Our work highlights glutamine anaplerosis as a potential therapeutic intervention point in MMA.
Collapse
Affiliation(s)
- Patrick Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ximena Bonilla
- Biomedical Informatics, Department of Computer Science, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | - David Lamparter
- Health 2030 Genome Center, Geneva, Switzerland
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
| | - Wenguang Shao
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Institute of Translational Medicine, Department of Health Science and Technology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | - Tanja Plessl
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Caroline Frei
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Bingisser
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sandra Goetze
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Institute of Translational Medicine, Department of Health Science and Technology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Audrey van Drogen
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Institute of Translational Medicine, Department of Health Science and Technology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | - Keith Harshman
- Health 2030 Genome Center, Geneva, Switzerland
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
| | - Patrick G A Pedrioli
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Institute of Translational Medicine, Department of Health Science and Technology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | | | - Martin Poms
- Division of Clinical Chemistry, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Florian Traversi
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Céline Bürer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sarah Cherkaoui
- Division of Oncology and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
| | - Raphael J Morscher
- Division of Oncology and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Luke Simmons
- Division of Child Neurology, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Merima Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ioannis Xenarios
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Agora Center, Lausanne, Switzerland
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | - Nicola Zamboni
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland
- Department of Biology, Institute of Molecular Systems Biology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland
| | - Gunnar Rätsch
- Biomedical Informatics, Department of Computer Science, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Medical Informatics Unit, University Hospital Zurich, Zurich, Switzerland.
- AI Center, ETH Zurich, Zurich, Switzerland.
| | - Emmanouil T Dermitzakis
- Health 2030 Genome Center, Geneva, Switzerland.
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland.
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland.
| | - Bernd Wollscheid
- PHRT Swiss Multi-Omics Center, smoc.ethz.ch, Zurich, Switzerland.
- Institute of Translational Medicine, Department of Health Science and Technology, Swiss Federal Institute of Technology/ETH Zürich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Starosta RT, Shinawi M. Primary Mitochondrial Disorders in the Neonate. Neoreviews 2022; 23:e796-e812. [PMID: 36450643 DOI: 10.1542/neo.23-12-e796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Primary mitochondrial disorders (PMDs) are a heterogeneous group of disorders characterized by functional or structural abnormalities in the mitochondria that lead to a disturbance of cellular energy, reactive oxygen species, and free radical production, as well as impairment of other intracellular metabolic functions, causing single- or multiorgan dysfunction. PMDs are caused by pathogenic variants in nuclear and mitochondrial genes, resulting in distinct modes of inheritance. Onset of disease is variable and can occur in the neonatal period, with a high morbidity and mortality. In this article, we review the most common methods used for the diagnosis of PMDs, as well as their prenatal and neonatal presentations. We highlight the shift in the diagnostic approach for PMDs since the introduction of nontargeted molecular tests into clinical practice, which has significantly reduced the use of invasive studies. We discuss common PMDs that can present in the neonate, including general, nonsyndromic presentations as well as specific syndromic disorders. We also review current treatment advances, including the use of mitochondrial "cocktails" based on limited scientific evidence and theoretical reasoning, as well as the impending arrival of personalized mitochondrial-specific treatments.
Collapse
Affiliation(s)
| | - Marwan Shinawi
- Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
32
|
Chinnery PF. Precision mitochondrial medicine. CAMBRIDGE PRISMS. PRECISION MEDICINE 2022; 1:e6. [PMID: 38550943 PMCID: PMC10953752 DOI: 10.1017/pcm.2022.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/29/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2024]
Abstract
Mitochondria play a key role in cell homeostasis as a major source of intracellular energy (adenosine triphosphate), and as metabolic hubs regulating many canonical cell processes. Mitochondrial dysfunction has been widely documented in many common diseases, and genetic studies point towards a causal role in the pathogenesis of specific late-onset disorder. Together this makes targeting mitochondrial genes an attractive strategy for precision medicine. However, the genetics of mitochondrial biogenesis is complex, with over 1,100 candidate genes found in two different genomes: the nuclear DNA and mitochondrial DNA (mtDNA). Here, we review the current evidence associating mitochondrial genetic variants with distinct clinical phenotypes, with some having clear therapeutic implications. The strongest evidence has emerged through the investigation of rare inherited mitochondrial disorders, but genome-wide association studies also implicate mtDNA variants in the risk of developing common diseases, opening to door for the incorporation of mitochondrial genetic variant analysis in population disease risk stratification.
Collapse
Affiliation(s)
- Patrick F. Chinnery
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
33
|
Macken WL, Falabella M, McKittrick C, Pizzamiglio C, Ellmers R, Eggleton K, Woodward CE, Patel Y, Labrum R, Phadke R, Reilly MM, DeVile C, Sarkozy A, Footitt E, Davison J, Rahman S, Houlden H, Bugiardini E, Quinlivan R, Hanna MG, Vandrovcova J, Pitceathly RDS. Specialist multidisciplinary input maximises rare disease diagnoses from whole genome sequencing. Nat Commun 2022; 13:6324. [PMID: 36344503 PMCID: PMC9640711 DOI: 10.1038/s41467-022-32908-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Diagnostic whole genome sequencing (WGS) is increasingly used in rare diseases. However, standard, semi-automated WGS analysis may overlook diagnoses in complex disorders. Here, we show that specialist multidisciplinary analysis of WGS, following an initial 'no primary findings' (NPF) report, improves diagnostic rates and alters management. We undertook WGS in 102 adults with diagnostically challenging primary mitochondrial disease phenotypes. NPF cases were reviewed by a genomic medicine team, thus enabling bespoke informatic approaches, co-ordinated phenotypic validation, and functional work. We enhanced the diagnostic rate from 16.7% to 31.4%, with management implications for all new diagnoses, and detected strong candidate disease-causing variants in a further 3.9% of patients. This approach presents a standardised model of care that supports mainstream clinicians and enhances diagnostic equity for complex disorders, thereby facilitating access to the potential benefits of genomic healthcare. This research was made possible through access to the data and findings generated by the 100,000 Genomes Project: http://www.genomicsengland.co.uk .
Collapse
Affiliation(s)
- William L Macken
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Caroline McKittrick
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Chiara Pizzamiglio
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Rebecca Ellmers
- Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Kelly Eggleton
- Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Cathy E Woodward
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Yogen Patel
- Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Robyn Labrum
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Neurogenetics Unit, Rare and Inherited Disease Laboratory, North Thames Genomic Laboratory Hub, London, UK
| | - Rahul Phadke
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Mary M Reilly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine DeVile
- Department of Neurosciences, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Emma Footitt
- Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - James Davison
- Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- National Institute for Health and Care Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Shamima Rahman
- Metabolic Unit, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Mitochondrial Research Group, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Enrico Bugiardini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Rosaline Quinlivan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Michael G Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Jana Vandrovcova
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK.
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK.
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
34
|
Wei W, Schon KR, Elgar G, Orioli A, Tanguy M, Giess A, Tischkowitz M, Caulfield MJ, Chinnery PF. Nuclear-embedded mitochondrial DNA sequences in 66,083 human genomes. Nature 2022; 611:105-114. [PMID: 36198798 PMCID: PMC9630118 DOI: 10.1038/s41586-022-05288-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/29/2022] [Indexed: 02/02/2023]
Abstract
DNA transfer from cytoplasmic organelles to the cell nucleus is a legacy of the endosymbiotic event-the majority of nuclear-mitochondrial segments (NUMTs) are thought to be ancient, preceding human speciation1-3. Here we analyse whole-genome sequences from 66,083 people-including 12,509 people with cancer-and demonstrate the ongoing transfer of mitochondrial DNA into the nucleus, contributing to a complex NUMT landscape. More than 99% of individuals had at least one of 1,637 different NUMTs, with 1 in 8 individuals having an ultra-rare NUMT that is present in less than 0.1% of the population. More than 90% of the extant NUMTs that we evaluated inserted into the nuclear genome after humans diverged from apes. Once embedded, the sequences were no longer under the evolutionary constraint seen within the mitochondrion, and NUMT-specific mutations had a different mutational signature to mitochondrial DNA. De novo NUMTs were observed in the germline once in every 104 births and once in every 103 cancers. NUMTs preferentially involved non-coding mitochondrial DNA, linking transcription and replication to their origin, with nuclear insertion involving multiple mechanisms including double-strand break repair associated with PR domain zinc-finger protein 9 (PRDM9) binding. The frequency of tumour-specific NUMTs differed between cancers, including a probably causal insertion in a myxoid liposarcoma. We found evidence of selection against NUMTs on the basis of size and genomic location, shaping a highly heterogenous and dynamic human NUMT landscape.
Collapse
Affiliation(s)
- Wei Wei
- Department of Clinical Neuroscience, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Katherine R Schon
- Department of Clinical Neuroscience, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
- Academic Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | - Marc Tischkowitz
- Academic Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Mark J Caulfield
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Patrick F Chinnery
- Department of Clinical Neuroscience, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
35
|
Hanaford AR, Cho YJ, Nakai H. AAV-vector based gene therapy for mitochondrial disease: progress and future perspectives. Orphanet J Rare Dis 2022; 17:217. [PMID: 35668433 PMCID: PMC9169410 DOI: 10.1186/s13023-022-02324-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/09/2022] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial diseases are a group of rare, heterogeneous diseases caused by gene mutations in both nuclear and mitochondrial genomes that result in defects in mitochondrial function. They are responsible for significant morbidity and mortality as they affect multiple organ systems and particularly those with high energy-utilizing tissues, such as the nervous system, skeletal muscle, and cardiac muscle. Virtually no effective treatments exist for these patients, despite the urgent need. As the majority of these conditions are monogenic and caused by mutations in nuclear genes, gene replacement is a highly attractive therapeutic strategy. Adeno-associated virus (AAV) is a well-characterized gene replacement vector, and its safety profile and ability to transduce quiescent cells nominates it as a potential gene therapy vehicle for several mitochondrial diseases. Indeed, AAV vector-based gene replacement is currently being explored in clinical trials for one mitochondrial disease (Leber hereditary optic neuropathy) and preclinical studies have been published investigating this strategy in other mitochondrial diseases. This review summarizes the preclinical findings of AAV vector-based gene replacement therapy for mitochondrial diseases including Leigh syndrome, Barth syndrome, ethylmalonic encephalopathy, and others.
Collapse
Affiliation(s)
- Allison R Hanaford
- Center for Integrative Brain Research, Seattle Children's Reserach Institute, Seattle, WA, 98101, USA.
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, 97239, USA.
| | - Yoon-Jae Cho
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR, 97239, USA
- Division of Pediatric Neurology, Doernbecher Children's Hospital, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Hiroyuki Nakai
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Molecular Immunology and Microbiology, Oregon Health and Science University, Portland, OR, 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| |
Collapse
|
36
|
Aldossary AM, Tawfik EA, Alomary MN, Alsudir SA, Alfahad AJ, Alshehri AA, Almughem FA, Mohammed RY, Alzaydi MM. Recent Advances in Mitochondrial Diseases: from Molecular Insights to Therapeutic Perspectives. Saudi Pharm J 2022; 30:1065-1078. [PMID: 36164575 PMCID: PMC9508646 DOI: 10.1016/j.jsps.2022.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022] Open
Abstract
Mitochondria are double-membraned cytoplasmic organelles that are responsible for the production of energy in eukaryotic cells. The process is completed through oxidative phosphorylation (OXPHOS) by the respiratory chain (RC) in mitochondria. Thousands of mitochondria may be present in each cell, depending on the function of that cell. Primary mitochondria disorder (PMD) is a clinically heterogeneous disease associated with germline mutations in mitochondrial DNA (mtDNA) and/or nuclear DNA (nDNA) genes, and impairs mitochondrial structure and function. Mitochondrial dysfunction can be detected in early childhood and may be severe, progressive and often multi-systemic, involving a wide range of organs. Understanding epigenetic factors and pathways mutations can help pave the way for developing an effective cure. However, the lack of information about the disease (including age of onset, symptoms, clinical phenotype, morbidity and mortality), the limits of current preclinical models and the wide range of phenotypic presentations hamper the development of effective medicines. Although new therapeutic approaches have been introduced with encouraging preclinical and clinical outcomes, there is no definitive cure for PMD. This review highlights recent advances, particularly in children, in terms of etiology, pathophysiology, clinical diagnosis, molecular pathways and epigenetic alterations. Current therapeutic approaches, future advances and proposed new therapeutic plans will also be discussed.
Collapse
|
37
|
A model to predict a risk of allergic rhinitis based on mitochondrial DNA copy number. Eur Arch Otorhinolaryngol 2022; 279:4997-5008. [DOI: 10.1007/s00405-022-07341-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/07/2022] [Indexed: 11/26/2022]
|
38
|
Schon K. Whole genome sequencing helps pinpoint a genetic diagnosis for patients. BMJ 2021; 375:n2680. [PMID: 34732386 DOI: 10.1136/bmj.n2680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Katherine Schon
- University of Cambridge & Cambridge University Hospitals NHS Trust
| |
Collapse
|
39
|
|